Skip to main content
Journal of Clinical and Translational Research logoLink to Journal of Clinical and Translational Research
. 2017 Feb 12;3(1):189–198. doi: 10.18053/jctres.03.2017S1.004

Connexin-based signaling and drug-induced hepatotoxicity

Michaël Maes, Mathieu Vinken 1
PMCID: PMC5562166  NIHMSID: NIHMS895086  PMID: 28825041

Abstract

Being critical mediators of liver homeostasis, connexins and their channels are frequently involved in liver toxicity. In the current paper, specific attention is paid to actions of hepatotoxic drugs on these communicative structures. In a first part, an overview is provided on the structural, regulatory and functional properties of connexin-based channels in the liver. In the second part, documented effects of acetaminophen, hypolipidemic drugs, phenobarbital and methapyriline on connexin signaling are discussed. Furthermore, the relevance of this subject for the fields of clinical and in vitro toxicology is demonstrated.

Relevance for patients: The role of connexin signaling in drug-induced hepatotoxicity may be of high clinical relevance, as it offers perspectives for the therapeutic treatment of such insults by interfering with connexin channel opening.

Keywords: gap junction, hemichannel, connexin, hepatotoxicity, drug-induced liver injury

1. Introduction

Gap junctions are goalkeepers of intercellular communication by mediating the passive diffusion of small and hydrophilic molecules, such as glutathione, adenosine triphosphate, cyclic adenosine monophosphate, inositol triphosphate, and ions, including calcium, sodium and potassium [1,2]. A plethora of physiological processes are regulated by substances that are intercellularly exchanged via gap junctions and hence gap junctional intercellular communication (GJIC) is considered as a key mechanism in the control of tissue homeostasis [3-13]. The liver was among the first organs in which gap junctions have been characterized [14,15]. More than 40 years ago, Goodenough isolated 2 gap junction proteins from mouse liver and called them connexins (Cx) [16]. At present, 21 different connexins have been identified in humans and rodents, all that are expressed in a cell type-specific way and named based on their molecular weight [17]. Nonetheless, they all share a common structure consisting of 4 transmembrane domains, 2 extracellular loops, 1 cytosolic loop, 1 cytosolic carboxyterminal tail and 1 cytosolic aminotail. Following synthesis, 6 connexins form a hemichannel at the plasma membrane surface, which then docks with another hemichannel from a neighboring cell to generate a gap junction [18-20] (Figure 1). This occurs at the extracellular domains, where conserved cysteine residues create disulfide bonds [21]. In recent years, it has become clear that undocked hemichannels may also provide a pathway for cellular signaling on their own independently of their role as structural precursors of gap junctions. Unlike their full channel counterparts, hemichannel communication occurs between an individual cell and its extracellular environment, yet the messengers that permeate hemichannels are very similar to those involved in GJIC [22-26]. Despite some structural variation between connexins, the first extra-cellular loop, the first transmembrane domain, the cytosolic aminotail and/or the cytosolic loop are considered to contribute to hemichannel pore opening [27]. Inherent to their participation in the maintenance of tissue homeostasis, connexins and their channels, in casu in liver, are also often involved in pathological processes, such as in liver disease and hepatotoxicity [28,29]. The present paper specifically focuses on the role of connexin signaling in drug-induced liver injury (DILI).

Figure 1. Structure of connexins and their channels.

Figure 1

Gap junctions group in so-called plaques at the plasma membrane surface and are formed by the docking of 2 hemichannels from neighboring cells, which in turn are built up by 6 connexins. Connexins share a similar structure consisting of 4 transmembrane domains (TM), 2 extracellular loops (EL), 1 cytosolic loop (CL), 1 cytosolic carboxyterminal tail (CT) and 1 cytosolic aminotail (NT).

2. Connexin-based channels in liver

2.1. Structural properties

Cx32 is the predominant connexin in liver and is expressed by hepatocytes and sinusoidal endothelial cells next to small quantities of Cx26, which is equally produced by stellate cells and Kupffer cells [30-32]. In addition, Cx43 is present in Kupffer cells, stellate cells, sinusoidal endothelial cells, cells of Glisson’s capsule and cholangiocytes [32-37], while Cx40 and Cx37 have been detected in liver vascular cells (Table 1) [38-40]. Nevertheless, functional gap junctions have thus far only been demonstrated in hepatocytes and stellate cells [32]. In fact, gap junctions in the pericentral and periportal acinar regions typically are Cx32 homotypic and Cx32-Cx26 heterotypic channels, respectively [35,41]. This complies with the observation that Cx26 is mainly expressed in the periportal area, whilst Cx32 is evenly distributed in liver tissue [42,43].

Table 1. Expression of connexins in liver.

Connexin Cell type References
Cx26 hepatocytes [164-166]
stellate cells [32]
sinusoidal endothelial cells [32]
Kupffer cells [32]
Cx32 hepatocytes [32,167]
biliary endothelial cells [36]
sinusoidal endothelial cells [32]
Cx37 hepatic artery endothelial cells [38-40]
portal vein endothelial cells [38-40]
Cx40 hepatic artery endothelial cells [38-40]
portal vein eindothelial cells [38-40]
Cx43 biliary epithelial cells [36, 168]
Kupffer cells [32-34,159]
stellate cells [32, 38]
sinusoidal enothelial cells [32, 38]
hepatic artery endothelial cells [38-40]
portal vein endothelials cells [38-40]

2.2. Regulatory properties

Connexin signaling can be regulated by a plethora of mechanisms at the transcriptional, posttranscriptional, translational and posttranslational level. As such, 2 major kinetic sources of regulation have been described, namely short-term control (i.e. millisecond to minute range) and long-term control (i.e. hour range). They cooperate to fine-tune the degree of intercellular communication by controlling the number of channels, their functional state and their unitary permeability [44,45].

Long-term control of GJIC involves regulation at the transcriptional level of connexin expression [44,46]. Connexin gene promoters show binding affinity for several ubiquitous transcription factors, such as activator protein 1. Furthermore, a number of cell type-specific transcription factors govern connexin gene transcription, including hepatocyte nuclear factor 1α that regulates Cx32 production in liver [47-49]. In addition, epigenetic mechanisms, in particular histone acetylation and DNA methylation, influence connexin gene expression [46,50], as shown in liver cells [51-53].

Short-term control of GJIC, so-called gating, is regulated by a variety of factors [54-57], among which posttranslational modifications, such as S-nitrosylation, sumoylation and phosphorylation, are prominent ones [58,59]. S-nitrosylation occurs at intracellular cysteine residues and is mediated by nitric oxide, which might be the underlying mechanism of increased hemichannel opening induced by metabolic inhibition and inflammatory conditions [60,61]. Irreversible conjugation of small ubiquitin-like modifiers to lysine residues, so-called sumoylation, regulates Cx43 levels and the number of Cx43-based gap junctions at the plasma membrane [62]. Phosphorylation encompasses the addition of phosphate groups to polar amino acid side chains, among which serine, threonine and tyrosine residues. This posttranslational modification almost uniquely takes place at the cytoplasmic carboxyterminal tail. With the exception of Cx26, all known connexins are phosphoproteins that are targeted by a broad panel of kinases. The regulation of gap junction opening by phosphorylation is complex and depends on the nature of the kinase and the identity of the connexin family member [55,59,63]. Cx43 may occur as a nonphosphorylated isoform and 2 phosphorylated isoforms [64-66]. In liver, Cx43 is mostly presented as the nonphosphorylated variant in quiescent conditions [52,67].

2.3. Functional properties

The establishment of GJIC is indispensable for the performance of many liver-specific functions, including albumin secretion [68], glycogenolysis [69-71], ammonia removal [68], bile secretion [72,73] and xenobiotic biotransformation [74-76]. Both the constitutive and drug-induced production of cytochrome P450 isoenzymes, in particular cytochrome P450 2B6 and 3A4, require the presence of Cx32-based gap junctions [77]. Induction of cytochrome P450 1A1/2 and 2B1/2 coincides with downregulation of pericentral Cx32 protein amounts in rat [74-76]. These concomitant changes may reflect a defense mechanism to restrict the intercellular diffusion of reactive intermediates produced through xenobiotic biotransformation [74]. Gap junctions composed of Cx32 also propagate glycogenolytic responses from the periportal to the peri-central pole, in particular by controlling the intercellular trafficking of inositol triphosphate [70]. The latter activates calcium release from endoplasmic reticulum stores, in turn evoking calcium waves throughout the acinar tract [78]. Likewise, bile secretion from cholangiocytes depends on the spread of calcium waves through Cx43-based gap junctions [36,73].

Upon partial hepatectomy, gap junction coupling intensifies in the G1 phase of the cell cycle, followed by a dramatic decrease during initiation of DNA synthesis. This is paralleled by similar changes in Cx32 expression [79-89]. In the regenerating liver of rats treated with an inhibitor of mitogen-activated protein kinase, the disappearance of Cx32 is inhibited without affecting hepatocyte proliferative activity [82], which suggests that downregulation of GJIC may occur independently of cell growth. However, in the regenerating liver of Cx32-/- mice, proliferative activity of the hepatocytes is not enhanced, yet the extent of synchronous initiation and termination of DNA synthesis is decreased. This may point to a supporting role for gap junctions in liver cell cycling [86,90]. The involvement of connexin signaling in liver cell growth may actually be more critical as anticipated. Thus, overexpression of Cx32 and Cx26 in rat liver epithelial cells and human hepatoma cells triggers the production of the cell cycle inhibitor p27 and the adherens junction protein E-cadherin, respectively, which, in turn, suppress proliferation [91].

Connexins and their channels have been reported to participate in different cell death processes in liver, including apoptosis [67,92,93], necrosis [94] and autophagy [95]. Interestingly, accumulating evidence suggests that connexin hemi-channels, rather than gap junctions, are involved in liver cell death. Following induction of apoptosis in primary hepatocytes, GJIC rapidly deteriorates, which is accompanied by a decay of the gap junctional Cx32 protein pool. Concomitantly, Cx32 is de novo synthesized and gathers in a hemichannel configuration. This becomes particularly evident towards the final stages of the cell death process, where Cx32 hemichannels facilitate the apoptosis-to-necrosis transition [92,96]. Along the same line, Cx43 signaling, also partly relying on hemichannels, was found to facilitate the onset of spontaneous apoptosis in cultures of primary hepatocytes [67].

3. Connexin-based channels and drug-induced liver injury

3.1. Acetaminophen

DILI is the leading cause of acute liver failure in Western countries with the vast majority being caused by overdosing with acetaminophen (APAP), a readily available analgesic and antipyretic drug [97,98]. After APAP intoxication in rodents, a switch in mRNA and protein production from Cx32 and Cx26 to Cx43 is observed [93,99]. The upregulation of Cx43 quantities is due to recruitment of Cx43-expressing inflammatory cells, but also originates from de novo production of hepatocytes [99]. In this regard, a recent study revealed that Cx43+/- mice display increased liver cell death, inflammation and oxidative stress in comparison with wild type (WT) littermates after APAP overdose [99]. These results suggest that newly synthesized hepatic Cx43 may protect against APAP-induced liver toxicity. A limited number of reports have described a role for Cx32-based gap junction in APAP-triggered hepatotoxicity using genetically modified animals, albeit with contradicting outcomes [93,100-102]. In this respect, Naiki-Ito and colleagues administered APAP to Cx32-dominant negative transgenic rats and noticed decreased aminotransferase serum levels and attenuated liver damage in comparison with WT animals [93]. Likewise, ceramide synthase 2-null mice, in which Cx32 is located in the cytosol of hepatocytes and that display aberrant GJIC, are less susceptible to APAP-induced hepatotoxicity [102]. In addition, an in vitro study showed protection against synchronized necrotic cell death of attached hepatocytes originating from Cx32+/+ mice compared to WT hepatocytes treated with APAP. This synchronization of cell death was mediated by gap junctions formed of Cx26 and Cx32. Furthermore, APAP-sensitive male hepatocytes were protected by attachment to APAP-insensitive female hepatocytes, with this protection being dependent on gap junctions. This points to a role for gap junction-based signaling in hepatocyte death by distribution of either death signaling molecules or survival messengers between hepatocytes [94]. In contrast, another report described increased serum amino-transferase levels and more pronounced liver insults in Cx32-/- mice after administration of APAP, indicating a cytoprotective function for hepatic Cx32 in APAP-induced injury, possibly linked to the trafficking of glutathione between hepatocytes via gap junctions [100]. This can be reconciled with the documented suppression of Cx32 production and simultaneous reduced channel activity upon exposure of hepatocytes to liver toxicants both in vitro and in vivo [29,101]. However, our group recently found that Cx32-/- mice form less protein adducts 6 hours after APAP administration, which could indicate a lower metabolic activity upon genetic ablation of Cx32 [101]. Indeed, at the more upstream mechanistic platform of APAP toxicity, cell death results from protein adduct formation involving N-acetyl-p-benzoquinone imine, the toxic metabolite of APAP [103,104]. This could question the suitability of genetically deficient rodents for investigating the role of Cx32 in APAP-induced hepatotoxicity. A possible alternative is the use of inhibitors of Cx32-based gap junctions. In this regard, a small molecule inhibitor of Cx32-based gap junctions, called 2-aminoethoxy-dipenyl-borate (2-APB), was reported to protect against liver failure and death in WT mice when co-administered with APAP [105]. However, a follow-up study demonstrated that the protection was only minor or completely lost when 2-APB was administered 1.5 hours or 4-6 hours, respectively, after APAP. In addition, part of the protection was due to the solvent dimethyl sulfoxide. Furthermore, in vitro experiments showed that the protection of 2-APB was caused by inhibition of metabolic activation of APAP as well as by inhibition of the c-jun-N-terminal kinase signaling pathway and not by blocking Cx32-based gap junctions [106]. In essence, de novo produced Cx43 after APAP-induced liver toxicity seems to have a protective role, while contradictory results were found with respect to the role of Cx32-based signaling.

3.2. Hypolipidemic drugs

Peroxisome proliferator-activated receptor α agonists, such as clofibrate [107], nafenopin [108] and Wy-14,643 [109] are lipid-lowering agents, which drive the expression of genes involved in fatty acid transport, binding and β-oxidation in favor of proliferative activity. Chronic treatment of rodents with peroxisome proliferators has been associated with hepatocarcinogenesis due to an induction of cell proliferation coupled to a suppression of hepatocyte apoptosis [107-109]. Both in vitro [110-112] and in vivo [113,114], it has been found that clofibrate, nafenopin and Wy-14,643 reduce hepatocellular GJIC. Inhibition of GJIC by Wy-14,643 occurs in a species-specific way, since it takes place in primary cultured hepatocytes from rat, mouse and hamster, but not from monkey and human [112]. Similarly, treatment of primary hepatocytes from rat, but not from guinea pig, with nafenopin causes reversible disappearance of GJIC [110]. The latter did not result from altered Cx26 and Cx32 protein levels or modifications in the cellular localization of Cx32, but was linked to protein kinase C-mediated phosphorylation of Cx32 [111]. By contrast, clofibrate [113,115] and Wy-14,643 [114] suppressed hepatic Cx26 and Cx32 protein levels. In addition, clofibrate enhanced the appearance of Cx43 in the cytoplasm of hepatocytes [113]. Overall, peroxisome proliferators seem to perturb GJIC and alter hepatic connexin expression. Stimulation of hepatocyte proliferation by these agents has also been shown to be mediated, at least in part, by tumor necrosis factor α (TNFα) [116,117]. Therefore, a conceivable explanation is that the downregulation of the connexin signaling is driven by TNFα released in response to peroxisome proliferators [114,118,119]. Indeed, TNFα treatment has been shown to modulate GJIC and to downregulate connexin gene expression [120]. Hence, GJIC inhibition by TNFα and subsequent promotion of hepatocyte proliferation might be a possible mechanistic interpretation of the effects of peroxisome proliferators in liver.

3.3. Phenobarbital

Phenobarbital or phenobarbitone (PB) is an anti-epileptic drug that has sedative and hypnotic properties. It is frequently used as a model tumor promoter in rodent liver and alters the expression of a broad set of genes [116,117], of which, those related to cytochrome P450-dependent xenobiotic biotransformation have gained most attention [118]. The presence of functional gap junctions consisting of Cx32, but not of Cx26, is a prerequisite for the promotional activity of PB, since Cx32-/- mice [121,122], unlike Cx26-/- mice [124], are resistant to promotion of hepatocarcinogenesis by this barbiturate. Further-more, a subset of genes is differentially affected by PB in the liver of Cx32-/- mice compared to their WT counterparts [123]. Interestingly, connexins are required for PB-mediated tumor promotion. It has been shown by several groups that gap junction activity becomes reduced upon administration of PB to rodents [74,113,125-128]. This is associated with abnormal frequency and size of gap junctions on the hepatocyte plasma membrane surface [129], decreased Cx32 immunoreactivity [74,125,130] and aberrant Cx32 localization [113,126], whereas Cx26 expression is not affected [74,125,126]. Both unchanged [74,128] and decreased [131,132] hepatic Cx32 mRNA levels are seen in PB-treated rodents. As shown in rodent models in vivo [128] and in vitro [133,134], the reduction of GJIC by PB occurs in a strain-specific way. Furthermore, the inhibitory effect of PB on GJIC between primary cultured mouse hepatocytes depends on xenobiotic biotransformation capacity, as it is abolished by a cytochrome P450 inhibitor [135].

3.4. Methapyrilene

Methapyrilene is an antihistamine with strong sedative properties that has been mainly prescribed to treat insomnia. It has been banned in most countries because of its potential to cause serious liver damage [136]. In recent years, methapyrilene has been tested in several toxicogenomics studies [136-140] and even in integrated systems toxicological trials [141] as a typical nongenotoxic hepatocarcinogen, whereby it became clear this drug induces numerous alterations in critical metabolic and signaling pathways. With respect to intercellular communication mediated by gap junctions, it has been found that the number and size of Cx32-containing gap junction plaques in liver are negatively affected upon treatment of male rats with a carcinogenic dose of methapyrilene. However, this dose also increased the occurrence of apoptosis, which may also contribute to the negative affect of methapyrilene on liver gap junctions [142].

4. Conclusions and perspectives

Because of its unique function and localization in the body, the liver is a primary target of toxicity induced by xenobiotics, including pharmaceuticals. Connexins and their channels are frequently involved in DILI, yet their exact role still is a matter of debate. In this light, Cx32-/- mice display lack of promotion of hepatocarcinogenesis by PB [121-123] and Wy-14,643 [143], suggesting that Cx32 signaling aggravates the adverse outcome. However, most evidence points to a rather defensive function for connexin signaling [90,144-149]. Thus, a high incidence of chemical-induced liver tumors was observed in mice deficient for Cx32 [90,144] and APAP-related liver injury is increased in Cx43+/- mice [99]. This discrepancy may be due, at least in part, to opposite actions of gap junctions and hemichannels. Indeed, while gap junctions are mainly associated with physiological functions, hemichannels are closed most of the time and seem to preferably open in pathological conditions [2,23,150,151]. Such differential effects of channels consisting of the same connexin building blocks are controversial and deserve further scrutiny. To add another layer of complexity, a novel class of connexin-like proteins has been identified in the last decade, namely the pannexins, which gather in a configuration identical to connexin hemichannels and that also provide an additional pathway for communication between the cytosol of individual cells and their extracellular environment [152,153]. Pannexins have been detected in a number of liver cells, in particular hepatocytes [154-159], and have been linked to lipoapoptosis [158]. Hence, pannexin signaling may also be potentially involved in drug-induced hepatotoxicity, a hypothesis that should be verified in the upcoming years.

The role of connexin signaling in DILI may be of high clinical relevance, as it offers perspectives for the therapeutic treatment of such insults by interfering with connexin channel opening. While doing is, care should be taken to develop specific channel modifiers. Besides the clinical toxicological importance, connexins and their channels are equally of interest to in vitro toxicologists. Specifically, inhibition of GJIC may represent a biomarker for the detection of nongenotoxic hepatocarcinogens, as shown for several drugs [114,142,160-163]. This could allow developing an in vitro assay for the testing of nongenotoxic carcinogenicity that might be used during early drug development [28].

Acknowledgments

This work was financially supported by grants from the European Research Council (ERC Starting Grant 335476), the Agency for Innovation by Science and Technology in Flanders (IWT grant 131003), the Fund for Scientific Research-Flanders (FWO grants G009514N and G010214N) and the University Hospital of the Vrije Universiteit Brussel-Belgium (“Willy Gepts Fonds” UZ-VUB).

Glossary

ABBREVIATIONS

Abbreviation Expansion
2-APB

2-aminoethoxy-dipenyl-borate

APAP

acetaminophen; Cx, connexin

DILI

drug-induced liver injury

GJIC

gap junctional intercellular communication

PB

phenobarbital

TNFα

tumor necrosis factor α

WT

wild type.

Disclosure

The authors report no conflict of interest.

References

  • [1].Alexander DB, Goldberg GS. Transfer of biologically important molecules between cells through gap junction channels. Curr Med Chem. 2003;10:2045–2058. doi: 10.2174/0929867033456927. [DOI] [PubMed] [Google Scholar]
  • [2].Decrock E, Vinken M, De Vuyst E, Krysko DV, D’Herde K, Vanhaecke T et al. Connexin-related signaling in cell death: to live or let die? Cell Death Differ. 2009;16:524–536. doi: 10.1038/cdd.2008.196. [DOI] [PubMed] [Google Scholar]
  • [3].Meens MJ, Kwak BR, Duffy HS. Role of connexins and pannexins in cardiovascular physiology. Cell Mol Life Sci. 2015;72:2779–2792. doi: 10.1007/s00018-015-1959-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Losa D, Chanson M. The lung communication network. Cell Mol Life Sci. 2015;72:2793–2808. doi: 10.1007/s00018-015-1960-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Maes M, Cogliati B, Crespo Yanguas S, Willebrords J, Vinken M. Roles of connexins and pannexins in digestive homeostasis. Cell Mol Life Sci. 2015;72:2809–2821. doi: 10.1007/s00018-015-1961-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Decrock E, De Bock M, Wang N, Bultynck G, Giaume C, Naus CC et al. Connexin and pannexin signaling pathways, an architectural blueprint for CNS physiology and pathology? Cell Mol Life Sci. 2015;72:2823–2851. doi: 10.1007/s00018-015-1962-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Plotkin LI, Stains JP. Connexins and pannexins in the skeleton: gap junctions, hemichannels and more. Cell Mol Life Sci. 2015;72:2853–2867. doi: 10.1007/s00018-015-1963-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Abed AB, Kavvadas P, Chadjichristos CE. Functional roles of connexins and pannexins in the kidney. Cell Mol Life Sci. 2015;72:2869–2877. doi: 10.1007/s00018-015-1964-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Kibschull M, Gellhaus A, Carette D, Segretain D, Pointis G, Gilleron J. Physiological roles of connexins and pannexins in reproductive organs. Cell Mol Life Sci. 2015;72:2879–2898. doi: 10.1007/s00018-015-1965-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Glass AM, Snyder EG, Taffet SM. Connexins and pannexins in the immune system and lymphatic organs. Cell Mol Life Sci. 2015;72:2899–2910. doi: 10.1007/s00018-015-1966-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Hodson DJ, Legros C, Desarménien MG, Guérineau NC. Roles of connexins and pannexins in (neuro)endocrine physiology. Cell Mol Life Sci. 2015;72:2911–2928. doi: 10.1007/s00018-015-1967-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Sáez JC, Cisterna BA, Vargas A, Cardozo CP. Regulation of pannexin and connexin channels and their functional role in skeletal muscles. Cell Mol Life Sci. 2015;72:2929–2935. doi: 10.1007/s00018-015-1968-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Faniku C, Wright CS, Martin PE. Connexins and pannexins in the integumentary system: the skin and appendages. Cell Mol Life Sci. 2015;72:2937–2947. doi: 10.1007/s00018-015-1969-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Loewenstein WR, Kanno Y. Intercellular communication and tissue growth. I. Cancerous growth. J Cell Biol. 1967;33:225–234. doi: 10.1083/jcb.33.2.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Revel JP, Karnovsky MJ. Hexagonal array of subunits in intercellular junctions of the mouse heart and liver. J Cell Biol. 1967;33:C7–C12. doi: 10.1083/jcb.33.3.c7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Goodenough DA. Bulk isolation of mouse hepatocyte gap junctions. Characterization of the principal protein, connexin. J Cell Biol. 1974;61:557–563. doi: 10.1083/jcb.61.2.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Söhl G, Willecke K. An update on connexin genes and their nomenclature in mouse and man. Cell Commun Adhes. 2003;10:173–180. doi: 10.1080/cac.10.4-6.173.180. [DOI] [PubMed] [Google Scholar]
  • [18].Dahl G, Werner R, Levine E, Rabadan-Diehl C. Mutational analysis of gap junction formation. Biophys J. 1992;62(172-180):180–172. doi: 10.1016/S0006-3495(92)81803-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Unger VM, Kumar NM, Gilula NB, Yeager M. Three-dimensional structure of a recombinant gap junction membrane channel. Science. 1999;283:1176–1180. doi: 10.1126/science.283.5405.1176. [DOI] [PubMed] [Google Scholar]
  • [20].Yeager M, Nicholson BJ. Structure of gap junction intercellular channels. Curr Opin Struct Biol. 1996;6:183–192. doi: 10.1016/s0959-440x(96)80073-x. [DOI] [PubMed] [Google Scholar]
  • [21].Foote CI, Zhou L, Zhu X, Nicholson BJ. The pattern of disulfide linkages in the extracellular loop regions of connexin 32 suggests a model for the docking interface of gap junctions. J Cell Biol. 1998;140:1187–1197. doi: 10.1083/jcb.140.5.1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Contreras JE, Sanchez HA, Eugenin EA, Speidel D, Theis M, Willecke K et al. Metabolic inhibition induces opening of unapposed connexin 43 gap junction hemichannels and reduces gap junctional communication in cortical astrocytes in culture. Proc Natl Acad Sci U S A. 2002;99:495–500. doi: 10.1073/pnas.012589799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Chandrasekhar A, Bera AK. Hemichannels: permeants and their effect on development, physiology and death. Cell Biochem Funct. 2012;30:89–100. doi: 10.1002/cbf.2794. [DOI] [PubMed] [Google Scholar]
  • [24].Spray DC, Ye ZC, Ransom BR. Functional connexin "hemichannels": a critical appraisal. Glia. 2006;54:758–773. doi: 10.1002/glia.20429. [DOI] [PubMed] [Google Scholar]
  • [25].Harris AL. Connexin channel permeability to cytoplasmic molecules. Prog Biophys Mol Biol. 2007;94:120–143. doi: 10.1016/j.pbiomolbio.2007.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Wang N, De Bock M, Decrock E, Bol M, Gadicherla A, Vinken M et al. Paracrine signaling through plasma membrane hemichannels. Biochim Biophys Acta. 2013;1828:35–50. doi: 10.1016/j.bbamem.2012.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Evans WH, De Vuyst E, Leybaert L. The gap junction cellular internet: connexin hemichannels enter the signalling limelight. Biochem J. 2006;397:1–14. doi: 10.1042/BJ20060175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Vinken M, Doktorova T, Decrock E, Leybaert L, Vanhaecke T, Rogiers V. Gap junctional intercellular communication as a target for liver toxicity and carcinogenicity. Crit Rev Biochem Mol Biol. 2009;44:201–222. doi: 10.1080/10409230903061215. [DOI] [PubMed] [Google Scholar]
  • [29].Vinken M. Gap junctions and non-neoplastic liver disease. J Hepatol. 2012;57:655–662. doi: 10.1016/j.jhep.2012.02.036. [DOI] [PubMed] [Google Scholar]
  • [30].Cascio M, Kumar NM, Safarik R, Gilula NB. Physical characterization of gap junction membrane connexons (hemi- channels) isolated from rat liver. J Biol Chem. 1995;270:18643–18648. doi: 10.1074/jbc.270.31.18643. [DOI] [PubMed] [Google Scholar]
  • [31].Neveu MJ, Hully JR, Babcock KL, Vaughan J, Hertzberg EL, Nicholson BJ et al. Proliferation-associated differences in the spatial and temporal expression of gap junction genes in rat liver. Hepatology. 1995;22:202–212. [PubMed] [Google Scholar]
  • [32].Fischer R, Reinehr R, Lu TP, Schonicke A, Warskulat U, Dienes HP et al. Intercellular communication via gap junctions in activated rat hepatic stellate cells. Gastroenterology. 2005;128:433–448. doi: 10.1053/j.gastro.2004.11.065. [DOI] [PubMed] [Google Scholar]
  • [33].Eugenin EA, Gonzalez HE, Sanchez HA, Branes MC, Saez JC. Inflammatory conditions induce gap junctional communication between rat Kupffer cells both in vivo and in vitro. Cell Immunol. 2007;247:103–110. doi: 10.1016/j.cellimm.2007.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Gonzalez HE, Eugenin EA, Garces G, Solis N, Pizarro M, Accatino L et al. Regulation of hepatic connexins in cholestasis: possible involvement of Kupffer cells and inflammatory mediators. Am J Physiol Gastrointest Liver Physiol. 2002;282:G991–G1001. doi: 10.1152/ajpgi.00298.2001. [DOI] [PubMed] [Google Scholar]
  • [35].Berthoud VM, Iwanij V, Garcia AM, Sáez JC. Connexins and glucagon receptors during development of rat hepatic acinus. Am J Physiol. 1992;263:650–658. doi: 10.1152/ajpgi.1992.263.5.G650. [DOI] [PubMed] [Google Scholar]
  • [36].Bode HP, Wang L, Cassio D, Leite MF, St-Pierre MV, Hirata K et al. Expression and regulation of gap junctions in rat cholangiocytes. Hepatology. 2002;36:631–640. doi: 10.1053/jhep.2002.35274. [DOI] [PubMed] [Google Scholar]
  • [37].Greenwel P, Rubin J, Schwartz M, Hertzberg EL, Rojkind M. Liver fat-storing cell clones obtained from a CCl4-cirrhotic rat are heterogeneous with regard to proliferation, expression of extracellular matrix components, interleukin-6, and connexin 43. Lab Invest. 1993;69:210–216. [PubMed] [Google Scholar]
  • [38].Hernández-Guerra M, González-Méndez Y, de Ganzo ZA, Salido E, García-Pagán JC, Abrante B et al. Role of gap junctions modulating hepatic vascular tone in cirrhosis. Liver Int. 2014;34:859–868. doi: 10.1111/liv.12446. [DOI] [PubMed] [Google Scholar]
  • [39].Shiojiri N, Niwa T, Sugiyama Y, Koike T. Preferential expression of connexin37 and connexin40 in the endothelium of the portal veins during mouse liver development. Cell Tissue Res. 2006;324:547–552. doi: 10.1007/s00441-006-0165-9. [DOI] [PubMed] [Google Scholar]
  • [40].Chaytor AT, Martin PE, Edwards DH, Griffith TM. Gap junctional communication underpins EDHF-type relaxations evoked by ACh in the rat hepatic artery. Am J Physiol Heart Circ Physiol. 2001;280:2441–2450. doi: 10.1152/ajpheart.2001.280.6.H2441. [DOI] [PubMed] [Google Scholar]
  • [41].Iwai M, Harada Y, Muramatsu A, Tanaka S, Mori T, Okanoue T et al. Development of gap junctional channels and intercellular communication in rat liver during ontogenesis. J Hepatol. 2000;32:11–18. doi: 10.1016/s0168-8278(00)80184-1. [DOI] [PubMed] [Google Scholar]
  • [42].Traub O, Look J, Dermietzel R, Brümmer F, Hülser D, Willecke K. Comparative characterization of the 21-kD and 26-kD gap junction proteins in murine liver and cultured hepatocytes. J Cell Biol. 1989;108:1039–1051. doi: 10.1083/jcb.108.3.1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Zhang JT, Nicholson BJ. The topological structure of connexin 26 and its distribution compared to connexin 32 in hepatic gap junctions. J Membr Biol. 1994;139:15–29. doi: 10.1007/BF00232671. [DOI] [PubMed] [Google Scholar]
  • [44].Oyamada M, Oyamada Y, Takamatsu T. Regulation of connexin expression. Biochim Biophys Acta. 2005;1719:6–23. doi: 10.1016/j.bbamem.2005.11.002. [DOI] [PubMed] [Google Scholar]
  • [45].Sáez JC, Retamal MA, Basilio D, Bukauskas FF, Bennett MV. Connexin-based gap junction hemichannels: gating mechanisms. Biochim Biophys Acta. 2005;1711:215–224. doi: 10.1016/j.bbamem.2005.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Oyamada M, Takebe K, Oyamada Y. Regulation of connexin expression by transcription factors and epigenetic mechanisms. Biochim Biophys Acta. 2013;1828:118–133. doi: 10.1016/j.bbamem.2011.12.031. [DOI] [PubMed] [Google Scholar]
  • [47].Piechocki MP, Toti RM, Fernstrom MJ, Burk RD, Ruch RJ. Liver cell-specific transcriptional regulation of connexin32. Biochim Biophys Acta. 2000;1491:107–122. doi: 10.1016/s0167-4781(00)00036-1. [DOI] [PubMed] [Google Scholar]
  • [48].Koffler LD, Fernstrom MJ, Akiyama TE, Gonzalez FJ, Ruch RJ. Positive regulation of connexin32 transcription by hepatocyte nuclear factor-1alpha. Arch Biochem Biophys. 2002;407:160–167. doi: 10.1016/s0003-9861(02)00488-5. [DOI] [PubMed] [Google Scholar]
  • [49].Field JM, Tate LA, Chipman JK, Minchin SD. Identification of functional regulatory regions of the connexin32 gene promoter. Biochim Biophys Acta. 2003;1628:22–29. doi: 10.1016/s0167-4781(03)00111-8. [DOI] [PubMed] [Google Scholar]
  • [50].Vinken M. Regulation of connexin signaling by the epigenetic machinery. Biochim Biophys Acta. 2016;1859:262–268. doi: 10.1016/j.bbagrm.2015.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Piechocki MP, Burk RD, Ruch RJ. Regulation of connexin32 and connexin43 gene expression by DNA methylation in rat liver cells. Carcinogenesis. 1999;20:401–406. doi: 10.1093/carcin/20.3.401. [DOI] [PubMed] [Google Scholar]
  • [52].Vinken M, Henkens T, Vanhaecke T, Papeleu P, Geerts A, Van Rossen E et al. Trichostatin a enhances gap junctional intercellular communication in primary cultures of adult rat hepatocytes. Toxicol Sci. 2006;91:484–492. doi: 10.1093/toxsci/kfj152. [DOI] [PubMed] [Google Scholar]
  • [53].Vinken M, Henkens T, Snykers S, Lukaszuk A, Tourwé D, Rogiers V et al. The novel histone deacetylase inhibitor 4-Me2N-BAVAH differentially affects cell junctions between primary hepatocytes. Toxicology. 2007;236:92–102. doi: 10.1016/j.tox.2007.04.003. [DOI] [PubMed] [Google Scholar]
  • [54].Bukauskas FF, Verselis VK. Gap junction channel gating. Biochim Biophys Acta. 2004;1662:42–60. doi: 10.1016/j.bbamem.2004.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Lampe PD, Lau AF. The effects of connexin phosphorylation on gap junctional communication. Int J Biochem Cell Biol. 2004;36:1171–1186. doi: 10.1016/S1357-2725(03)00264-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Peracchia C. Chemical gating of gap junction channels; roles of calcium, pH and calmodulin. Biochim Biophys Acta. 2004;1662:61–80. doi: 10.1016/j.bbamem.2003.10.020. [DOI] [PubMed] [Google Scholar]
  • [57].Cottrell GT, Burt JM. Functional consequences of heterogeneous gap junction channel formation and its influence in health and disease. Biochim Biophys Acta. 2005;1711:126–141. doi: 10.1016/j.bbamem.2004.11.013. [DOI] [PubMed] [Google Scholar]
  • [58].Pogoda K, Kameritsch P, Retamal MA, Vega JL. Regulation of gap junction channels and hemichannels by phosphorylation and redox changes: a revision. BMC Cell Biol. 2016;17(1):11. doi: 10.1186/s12860-016-0099-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Johnstone SR, Billaud M, Lohman AW, Taddeo EP, Isakson BE. Posttranslational modifications in connexins and pannexins. J Membr Biol. 2012;245:319–332. doi: 10.1007/s00232-012-9453-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Retamal MA, Cortés CJ, Reuss L, Bennett MV, Sáez JC. S-nitrosylation and permeation through connexin 43 hemi-channels in astrocytes: induction by oxidant stress and reversal by reducing agents. Proc Natl Acad Sci U S A. 2006;103:4475–4480. doi: 10.1073/pnas.0511118103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Retamal MA, Froger N, Palacios-Prado N, Ezan P, Saez PJ, Saez JC et al. Cx43 hemichannels and gap junction channels in astrocytes are regulated oppositely by proinflammatory cytokines released from activated microglia. J Neurosci. 2007;27:13781–13792. doi: 10.1523/JNEUROSCI.2042-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Kjenseth A, Fykerud TA, Sirnes S, Bruun J, Yohannes Z, Kolberg M et al. The gap junction channel protein connexin 43 is covalently modified and regulated by SUMOylation. J Biol Chem. 2012;287:15851–15861. doi: 10.1074/jbc.M111.281832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Laird DW. Connexin phosphorylation as a regulatory event linked to gap junction internalization and degradation. Biochim Biophys Acta. 2005;1711:172–182. doi: 10.1016/j.bbamem.2004.09.009. [DOI] [PubMed] [Google Scholar]
  • [64].Cooper CD, Solan JL, Dolejsi MK, Lampe PD. Analysis of connexin phosphorylation sites. Methods. 2000;20:196–204. doi: 10.1006/meth.1999.0937. [DOI] [PubMed] [Google Scholar]
  • [65].Solan JL, Lampe PD. Connexin43 phosphorylation: structural changes and biological effects. Biochem J. 2009;419:261–272. doi: 10.1042/BJ20082319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Solan JL, Lampe PD. Connexin phosphorylation as a regulatory event linked to gap junction channel assembly. Biochim Biophys Acta. 2005;1711:154–163. doi: 10.1016/j.bbamem.2004.09.013. [DOI] [PubMed] [Google Scholar]
  • [67].Vinken M, Decrock E, Vanhaecke T, Leybaert L, Rogiers V. Connexin43 signaling contributes to spontaneous apoptosis in cultures of primary hepatocytes. Toxicol Sci. 2012;125:175–186. doi: 10.1093/toxsci/kfr277. [DOI] [PubMed] [Google Scholar]
  • [68].Yang J, Ichikawa A, Tsuchiya T. A novel function of connexin 32: marked enhancement of liver function in a hepatoma cell line. Biochem Biophys Res Commun. 2003;307:80–85. doi: 10.1016/s0006-291x(03)01117-3. [DOI] [PubMed] [Google Scholar]
  • [69].Stumpel F, Ott T, Willecke K, Jungermann K. Connexin 32 gap junctions enhance stimulation of glucose output by glucagon and noradrenaline in mouse liver. Hepatology. 1998;28:1616–1620. doi: 10.1002/hep.510280622. [DOI] [PubMed] [Google Scholar]
  • [70].Clair C, Chalumeau C, Tordjmann T, Poggioli J, Erneux C, Dupont G et al. Investigation of the roles of Ca(2+) and InsP(3) diffusion in the coordination of Ca(2+) signals between connected hepatocytes. J Cell Sci. 2001;114:1999–2007. doi: 10.1242/jcs.114.11.1999. [DOI] [PubMed] [Google Scholar]
  • [71].Nelles E, Butzler C, Jung D, Temme A, Gabriel HD, Dahl U et al. Defective propagation of signals generated by sympathetic nerve stimulation in the liver of connexin32-deficient mice. Proc Natl Acad Sci U S A. 1996;93:9565–9570. doi: 10.1073/pnas.93.18.9565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Temme A, Stumpel F, Sohl G, Rieber EP, Jungermann K, Willecke K et al. Dilated bile canaliculi and attenuated decrease of nerve-dependent bile secretion in connexin32- deficient mouse liver. Pflugers Archiv. 2001;442:961–966. doi: 10.1007/s004240100623. [DOI] [PubMed] [Google Scholar]
  • [73].Nathanson MH, Rios-Velez L, Burgstahler AD, Mennone A. Communication via gap junctions modulates bile secretion in the isolated perfused rat liver. Gastroenterology. 1999;116:1176–1183. doi: 10.1016/s0016-5085(99)70021-1. [DOI] [PubMed] [Google Scholar]
  • [74].Neveu MJ, Babcock KL, Hertzberg EL, Paul DL, Nicholson BJ, Pitot HC. Colocalized alterations in connexin32 and cytochrome P450IIB1/2 by phenobarbital and related liver tumor promoters. Cancer Res. 1994;54:3145–3152. [PubMed] [Google Scholar]
  • [75].Shoda T, Mitsumori K, Onodera H, Toyoda K, Uneyama C, Imazawa T et al. The relationship between decrease in Cx32 and induction of P450 isozymes in the early phase of clofibrate hepatocarcinogenesis in the rat. Arch Toxicol. 1999;73:373–380. doi: 10.1007/s002040050676. [DOI] [PubMed] [Google Scholar]
  • [76].Shoda T, Mitsumori K, Onodera H, Toyoda K, Uneyama C, Takada K et al. Liver tumor-promoting effect of beta- naphthoflavone, a strong CYP 1A1/2 inducer, and the relationship between CYP 1A1/2 induction and Cx32 decrease in its hepatocarcinogenesis in the rat. Toxicol Pathol. 2000;28:540–547. doi: 10.1177/019262330002800406. [DOI] [PubMed] [Google Scholar]
  • [77].Hamilton GA, Jolley SL, Gilbert D, Coon DJ, Barros S, LeCluyse EL. Regulation of cell morphology and cytochrome P450 expression in human hepatocytes by extracellular matrix and cell-cell interactions. Cell Tissue Res. 2001;306:85–99. doi: 10.1007/s004410100429. [DOI] [PubMed] [Google Scholar]
  • [78].Gaspers LD, Thomas AP. Calcium signaling in liver. Cell Calcium. 2005;38:329–342. doi: 10.1016/j.ceca.2005.06.009. [DOI] [PubMed] [Google Scholar]
  • [79].Dermietzel R, Yancey SB, Traub O, Willecke K, Revel JP. Major loss of the 28-kD protein of gap junction in proliferating hepatocytes. J Cell Biol. 1987;105:1925–1934. doi: 10.1083/jcb.105.4.1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Fladmark KE, Gjertsen BT, Molven A, Mellgren G, Vintermyr OK, Døskeland SO. Gap junctions and growth control in liver regeneration and in isolated rat hepatocytes. Hepatology. 1997;25:847–855. doi: 10.1002/hep.510250411. [DOI] [PubMed] [Google Scholar]
  • [81].Koenig S, Krause P, Drabent B, Schaeffner I, Christ B, Schwartz P et al. The expression of mesenchymal, neural and haematopoietic stem cell markers in adult hepatocytes proliferating in vitro. J Hepatol. 2006;44:1115–1124. doi: 10.1016/j.jhep.2005.09.016. [DOI] [PubMed] [Google Scholar]
  • [82].Kojima T, Yamamoto T, Murata M, Lan M, Takano K, Go M et al. Role of the p38 MAP-kinase signaling pathway for Cx32 and claudin-1 in the rat liver. Cell Commun Adhes. 2003;10:437–443. doi: 10.1080/cac.10.4-6.437.443. [DOI] [PubMed] [Google Scholar]
  • [83].Kren BT, Kumar NM, Wang SQ, Gilula NB, Steer CJ. Differential regulation of multiple gap junction transcripts and proteins during rat liver regeneration. J Cell Biol. 1993;123:707–718. doi: 10.1083/jcb.123.3.707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Meyer DJ, Yancey SB, Revel JP. Intercellular communication in normal and regenerating rat liver: a quantitative analysis. J Cell Biol. 1981;91:505–523. doi: 10.1083/jcb.91.2.505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Miyashita T, Takeda A, Iwai M, Shimazu T. Single administration of hepatotoxic chemicals transiently decreases the gap-junction-protein levels of connexin 32 in rat liver. Eur J Biochem. 1991;196:37–42. doi: 10.1111/j.1432-1033.1991.tb15782.x. [DOI] [PubMed] [Google Scholar]
  • [86].Sugiyama Y, Ohta H. Changes in density and distribution of gap junctions after partial hepatectomy: immunohistochemical and morphometric studies. Arch Histol Cytol. 1990;53:71–80. doi: 10.1679/aohc.53.71. [DOI] [PubMed] [Google Scholar]
  • [87].Temme A, Ott T, Dombrowski F, Willecke K. The extent of synchronous initiation and termination of DNA synthesis in regenerating mouse liver is dependent on connexin32 expressing gap junctions. J Hepatol. 2000;32:627–635. doi: 10.1016/s0168-8278(00)80225-1. [DOI] [PubMed] [Google Scholar]
  • [88].Traub O, Drüge PM, Willecke K. Degradation and resynthesis of gap junction protein in plasma membranes of regenerating liver after partial hepatectomy or cholestasis. Proc Natl Acad Sci U S A. 1983;80:755–759. doi: 10.1073/pnas.80.3.755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Yee AG, Revel JP. Loss and reappearance of gap junctions in regenerating liver. J Cell Biol. 1978;78:554–564. doi: 10.1083/jcb.78.2.554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Dagli ML, Yamasaki H, Krutovskikh V, Omori Y. Delayed liver regeneration and increased susceptibility to chemical hepatocarcinogenesis in transgenic mice expressing a dominant-negative mutant of connexin32 only in the liver. Carcinogenesis. 2004;25:483–492. doi: 10.1093/carcin/bgh050. [DOI] [PubMed] [Google Scholar]
  • [91].Paul DL. Molecular cloning of cDNA for rat liver gap junction protein. J Cell Biol. 1986;103:123–134. doi: 10.1083/jcb.103.1.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Vinken M, Decrock E, De Vuyst E, De Bock M, Vandenbroucke RE, De Geest BG et al. Connexin32 hemichannels contribute to the apoptotic-to-necrotic transition during Fas-mediated hepatocyte cell death. Cell Mol Life Sci. 2010;67:907–918. doi: 10.1007/s00018-009-0220-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Naiki-Ito A, Asamoto M, Naiki T, Ogawa K, Takahashi S, Sato S et al. Gap junction dysfunction reduces acetaminophen hepatotoxicity with impact on apoptotic signaling and connexin 43 protein induction in rat. Toxicol Pathol. 2010;38:280–286. doi: 10.1177/0192623309357951. [DOI] [PubMed] [Google Scholar]
  • [94].Saito C, Shinzawa K, Tsujimoto Y. Synchronized necrotic death of attached hepatocytes mediated via gap junctions. Sci Rep. 2014;4:5169. doi: 10.1038/srep05169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Zou H, Zhuo L, Han T, Hu D, Yang X, Wang Y et al. Autophagy and gap junctional intercellular communication inhibition are involved in cadmium-induced apoptosis in rat liver cells. Biochem Biophys Res Commun. 2015;459:713–719. doi: 10.1016/j.bbrc.2015.03.027. [DOI] [PubMed] [Google Scholar]
  • [96].Kalvelyte A, Imbrasaite A, Bukauskiene A, Verselis VK, Bukauskas FF. Connexins and apoptotic transformation. Biochem Pharmacol. 2003;66:1661–1672. doi: 10.1016/s0006-2952(03)00540-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Ichai P, Samuel D. Epidemiology of liver failure. Clin Res Hepatol Gastroenterol. 2011;35:610–617. doi: 10.1016/j.clinre.2011.03.010. [DOI] [PubMed] [Google Scholar]
  • [98].Lee WM. Etiologies of acute liver failure. Semin Liver Dis. 2008;28:142–152. doi: 10.1055/s-2008-1073114. [DOI] [PubMed] [Google Scholar]
  • [99].Maes M, McGill MR, da Silva TC, Abels C, Lebofsky M, de Araujo CMM et al. Involvement of connexin43 in acetaminophen-induced liver injury. Biochim Biophys Acta. 2016;1862:1111–1121. doi: 10.1016/j.bbadis.2016.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Igarashi I, Maejima T, Kai K, Arakawa S, Teranishi M, Sanbuissho A. Role of connexin 32 in acetaminophen toxicity in a knockout mice model. Exp Toxicol Pathol. 2014;66:103–110. doi: 10.1016/j.etp.2013.10.002. [DOI] [PubMed] [Google Scholar]
  • [101].Maes M, McGill MR, da Silva TC, Lebofsky M, de Araujo CMM, Tiburcio T et al. Connexin32: a mediator of acetaminophen-induced liver injury? Toxicol Mech Methods. 2016;26:88–96. doi: 10.3109/15376516.2015.1103000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Park WJ, Park JW, Erez-Roman R, Kogot-Levin A, Bame JR, Tirosh B et al. Protection of a ceramide synthase 2 null mouse from drug-induced liver injury: role of gap junction dysfunction and connexin 32 mislocalization. J Biol Chem. 2013;288:30904–30916. doi: 10.1074/jbc.M112.448852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Dahlin DC, Miwa GT, Lu AY, Nelson SD. N-acetyl-p-benzoquinone imine: a cytochrome P-450-mediated oxidation product of acetaminophen. Proc Natl Acad Sci U S A. 1984;81:1327–1331. doi: 10.1073/pnas.81.5.1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Maes M, Vinken M, Jaeschke H. Experimental models of hepatotoxicity related to acute liver failure. Toxicol Appl Pharmacol. 2016;290:86–97. doi: 10.1016/j.taap.2015.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Patel SJ, Milwid JM, King KR, Bohr S, Iracheta-Velle A, Li M et al. Gap junction inhibition prevents drug-induced liver toxicity and fulminant hepatic failure. Nat Biotechnol. 2012;30:179–183. doi: 10.1038/nbt.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Du K, Williams CD, McGill MR, Xie Y, Farhood A, Vinken M et al. The gap junction inhibitor 2-aminoethoxy-diphenyl- borate protects against acetaminophen hepatotoxicity by inhibiting cytochrome P450 enzymes and c-jun N-terminal kinase activation. Toxicol Appl Pharmacol. 2013;273:484–491. doi: 10.1016/j.taap.2013.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Fidaleo M. Human health risk assessment for peroxisome proliferators: more than 30 years of research. Exp Toxicol Pathol. 2009;61:215–221. doi: 10.1016/j.etp.2008.09.002. [DOI] [PubMed] [Google Scholar]
  • [108].Roberts RA, Chevalier S, Hasmall SC, James NH, Cosulich SC, Macdonald N. PPAR alpha and the regulation of cell division and apoptosis. Toxicology. 2002;181:167–170. doi: 10.1016/s0300-483x(02)00275-5. [DOI] [PubMed] [Google Scholar]
  • [109].Gonzalez FJ, Shah YM. PPARalpha: mechanism of species differences and hepatocarcinogenesis of peroxisome proliferators. Toxicology. 2008;246:2–8. doi: 10.1016/j.tox.2007.09.030. [DOI] [PubMed] [Google Scholar]
  • [110].Elcock FJ, Chipman JK, Roberts RA. The rodent nongenotoxic hepatocarcinogen and peroxisome proliferator nafenopin inhibits intercellular communication in rat but not guineapig hepatocytes, perturbing S-phase but not apoptosis. Arch Toxicol. 1998;72:439–444. doi: 10.1007/s002040050524. [DOI] [PubMed] [Google Scholar]
  • [111].Elcock FJ, Deag E, Roberts RA, Chipman JK. Nafenopin causes protein kinase C-mediated serine phosphorylation and loss of function of connexin 32 protein in rat hepatocytes without aberrant expression or localization. Toxicol Sci. 2000;56:86–94. doi: 10.1093/toxsci/56.1.86. [DOI] [PubMed] [Google Scholar]
  • [112].Kamendulis LM, Isenberg JS, Smith JH, Pugh G, Lington AW, Klaunig JE. Comparative effects of phthalate monoesters on gap junctional intercellular communication and peroxisome proliferation in rodent and primate hepatocytes. J Toxicol Environ Health A. 2002;65:569–588. doi: 10.1080/152873902317349736. [DOI] [PubMed] [Google Scholar]
  • [113].Krutovskikh VA, Mesnil M, Mazzoleni G, Yamasaki H. Inhibition of rat liver gap junction intercellular communication by tumor-promoting agents in vivo. Association with aberrant localization of connexin proteins. Lab Invest. 1995;72:571–577. [PubMed] [Google Scholar]
  • [114].Cowles C, Mally A, Chipman JK. Different mechanisms of modulation of gap junction communication by non-genotoxic carcinogens in rat liver in vivo. Toxicology. 2007;238:49–59. doi: 10.1016/j.tox.2007.05.027. [DOI] [PubMed] [Google Scholar]
  • [115].Tsuda H, Asamoto M, Baba-Toriyama H, Iwahori Y, Hori T, Kim DJ et al. Clofibrate-induced neoplastic development in the rat liver is associated with decreased connexin 32 expression but not with a co-ordinated shift in expression of marker enzymes. Toxicol Lett. 1995;82-83:693–699. doi: 10.1016/0378-4274(95)03587-7. [DOI] [PubMed] [Google Scholar]
  • [116].Bojes HK, Germolec DR, Simeonova P, Bruccoleri A, Schoonhoven R, Luster MI et al. Antibodies to tumor necrosis factor alpha prevent increases in cell replication in liver due to the potent peroxisome proliferator, WY-14,643. Carcinogenesis. 1997;18:669–674. doi: 10.1093/carcin/18.4.669. [DOI] [PubMed] [Google Scholar]
  • [117].Roberts RA, James NH, Cosulich S, Hasmall SC, Orphanides G. Role of cytokines in non-genotoxic hepatocarcinogenesis: cause or effect? Toxicol Lett. 2001;120:301–306. doi: 10.1016/s0378-4274(01)00282-x. [DOI] [PubMed] [Google Scholar]
  • [118].Chandross KJ, Spray DC, Cohen RI, Kumar NM, Kremer M, Dermietzel R et al. TNF alpha inhibits Schwann cell proliferation, connexin46 expression, and gap junctional communication. Mol Cell Neurosci. 1996;7:479–500. doi: 10.1006/mcne.1996.0035. [DOI] [PubMed] [Google Scholar]
  • [119].van Rijen HV, van Kempen MJ, Postma S, Jongsma HJ. Tumour necrosis factor alpha alters the expression of connexin43, connexin40, and connexin37 in human umbilical vein endothelial cells. Cytokine. 1998;10:258–264. doi: 10.1006/cyto.1997.0287. [DOI] [PubMed] [Google Scholar]
  • [120].Mensink A, de Haan LH, Lakemond CM, Koelman CA, Koeman JH. Inhibition of gap junctional intercellular communication between primary human smooth muscle cells by tumor necrosis factor alpha. Carcinogenesis. 1995;16:2063–2067. doi: 10.1093/carcin/16.9.2063. [DOI] [PubMed] [Google Scholar]
  • [121].Moennikes O, Buchmann A, Romualdi A, Ott T, Werringloer J, Willecke K et al. Lack of phenobarbital-mediated promotion of hepatocarcinogenesis in connexin32-null mice. Cancer Res. 2000;60:5087–5091. [PubMed] [Google Scholar]
  • [122].Luebeck EG, Buchmann A, Schneider D, Moolgavkar SH, Schwarz M. Modulation of liver tumorigenesis in Connexin32-deficient mouse. Mutat Res. 2005;570:33–47. doi: 10.1016/j.mrfmmm.2004.09.008. [DOI] [PubMed] [Google Scholar]
  • [123].Stahl S, Ittrich C, Marx-Stoelting P, Köhle C, Ott T, Buchmann A et al. Effect of the tumor promoter phenobarbital on the pattern of global gene expression in liver of connexin32- wild-type and connexin32-deficient mice. Int J Cancer. 2005;115:861–869. doi: 10.1002/ijc.20815. [DOI] [PubMed] [Google Scholar]
  • [124].Marx-Stoelting P, Mahr J, Knorpp T, Schreiber S, Templin MF, Ott T et al. Tumor promotion in liver of mice with a conditional Cx26 knockout. Toxicol Sci. 2008;103:260–267. doi: 10.1093/toxsci/kfn043. [DOI] [PubMed] [Google Scholar]
  • [125].Neveu MJ, Hully JR, Paul DL, Pitot HC. Reversible alteration in the expression of the gap junctional protein connexin 32 during tumor promotion in rat liver and its role during cell proliferation. Cancer Commun. 1990;2:21–31. doi: 10.3727/095535490820874731. [DOI] [PubMed] [Google Scholar]
  • [126].Ito S, Tsuda M, Yoshitake A, Yanai T, Masegi T. Effect of phenobarbital on hepatic gap junctional intercellular communication in rats. Toxicol Pathol. 1998;26:253–259. doi: 10.1177/019262339802600210. [DOI] [PubMed] [Google Scholar]
  • [127].Jeong SH, Habeebu SS, Klaassen CD. Cadmium decreases gap junctional intercellular communication in mouse liver. Toxicol Sci. 2000;57:156–166. doi: 10.1093/toxsci/57.1.156. [DOI] [PubMed] [Google Scholar]
  • [128].Warner KA, Fernstrom MJ, Ruch RJ. Inhibition of mouse hepatocyte gap junctional intercellular communication by phenobarbital correlates with strain-specific hepatocarcinogenesis. Toxicol Sci. 2003;71:190–197. doi: 10.1093/toxsci/71.2.190. [DOI] [PubMed] [Google Scholar]
  • [129].Sugie S, Mori H, Takahashi M. Effect of in vivo exposure to the liver tumor promoters phenobarbital or DDT on the gap junctions of rat hepatocytes: a quantitative freeze-fracture analysis. Carcinogenesis. 1987;8:45–51. doi: 10.1093/carcin/8.1.45. [DOI] [PubMed] [Google Scholar]
  • [130].Okamiya H, Mitsumori K, Onodera H, Ito S, Imazawa T, Yasuhara K et al. Mechanistic study on liver tumor promoting effects of piperonyl butoxide in rats. Arch Toxicol. 1998;72:744–750. doi: 10.1007/s002040050569. [DOI] [PubMed] [Google Scholar]
  • [131].Beer DG, Neveu MJ, Paul DL, Rapp UR, Pitot HC. Expression of the c-raf protooncogene, gamma-glutamyltranspeptidase, and gap junction protein in rat liver neoplasms. Cancer Res. 1988;48:1610–1617. [PubMed] [Google Scholar]
  • [132].Mesnil M, Fitzgerald DJ, Yamasaki H. Phenobarbital specifically reduces gap junction protein mRNA level in rat liver. Mol Carcinog. 1988;1:79–81. doi: 10.1002/mc.2940010202. [DOI] [PubMed] [Google Scholar]
  • [133].Klaunig JE, Ruch RJ. Strain and species effects on the inhibition of hepatocyte intercellular communication by liver tumor promoters. Cancer Lett. 1987;36:161–168. doi: 10.1016/0304-3835(87)90087-5. [DOI] [PubMed] [Google Scholar]
  • [134].Ren P, Mehta PP, Ruch RJ. Inhibition of gap junctional intercellular communication by tumor promoters in connexin43 and connexin32-expressing liver cells: cell specificity and role of protein kinase C. Carcinogenesis. 1998;19:169–175. doi: 10.1093/carcin/19.1.169. [DOI] [PubMed] [Google Scholar]
  • [135].Klaunig JE, Ruch RJ, Weghorst CM. Comparative effects of phenobarbital, DDT, and lindane on mouse hepatocyte gap junctional intercellular communication. Toxicol Appl Pharmacol. 1990;102:553–563. doi: 10.1016/0041-008x(90)90050-5. [DOI] [PubMed] [Google Scholar]
  • [136].Auman JT, Chou J, Gerrish K, Huang Q, Jayadev S, Blanchard K et al. Identification of genes implicated in methapyrilene- induced hepatotoxicity by comparing differential gene expression in target and nontarget tissue. Environ Health Perspect. 2007;115:572–578. doi: 10.1289/ehp.9396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Hamadeh HK, Knight BL, Haugen AC, Sieber S, Amin RP, Bushel PR et al. Methapyrilene toxicity: anchorage of pathologic observations to gene expression alterations. Toxicol Pathol. 2002;30:470–482. doi: 10.1080/01926230290105712. [DOI] [PubMed] [Google Scholar]
  • [138].Waring JF, Ulrich RG, Flint N, Morfitt D, Kalkuhl A, Staedtler F et al. Interlaboratory evaluation of rat hepatic gene expression changes induced by methapyrilene. Environ Health Perspect. 2004;112:439–448. doi: 10.1289/ehp.6643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Beekman JM, Boess F, Hildebrand H, Kalkuhl A, Suter L. Gene expression analysis of the hepatotoxicant methapyrilene in primary rat hepatocytes: an interlaboratory study. Environ Health Perspect. 2006;114:92–99. doi: 10.1289/ehp.7915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Uehara T, Kiyosawa N, Hirode M, Omura K, Shimizu T, Ono A et al. Gene expression profiling of methapyrilene-induced hepatotoxicity in rat. J Toxicol Sci. 2008;33:37–50. doi: 10.2131/jts.33.37. [DOI] [PubMed] [Google Scholar]
  • [141].Craig A, Sidaway J, Holmes E, Orton T, Jackson D, Rowlinson R et al. Systems toxicology: integrated genomic, proteomic and metabonomic analysis of methapyrilene induced hepatotoxicity in the rat. J Proteome Res. 2006;5:1586–1601. doi: 10.1021/pr0503376. [DOI] [PubMed] [Google Scholar]
  • [142].Mally A, Chipman JK. Non-genotoxic carcinogens: early effects on gap junctions, cell proliferation and apoptosis in the rat. Toxicology. 2002;180:233–248. doi: 10.1016/s0300-483x(02)00393-1. [DOI] [PubMed] [Google Scholar]
  • [143].Moennikes O, Stahl S, Bannasch P, Buchmann A, Schwarz M. WY-14, 643-mediated promotion of hepatocarcinogenesis in connexin32-wild-type and connexin32-null mice. Carcinogenesis. 2003;24:1561–1565. doi: 10.1093/carcin/bgg099. [DOI] [PubMed] [Google Scholar]
  • [144].Temme A, Buchmann A, Gabriel HD, Nelles E, Schwarz M, Willecke K. High incidence of spontaneous and chemically induced liver tumors in mice deficient for connexin32. Curr Biol. 1997;7:713–716. doi: 10.1016/s0960-9822(06)00302-2. [DOI] [PubMed] [Google Scholar]
  • [145].King TJ, Lampe PD. Mice deficient for the gap junction protein Connexin32 exhibit increased radiation-induced tumorigenesis associated with elevated mitogen-activated protein kinase (p44/Erk1, p42/Erk2) activation. Carcinogenesis. 2004;25:669–680. doi: 10.1093/carcin/bgh071. [DOI] [PubMed] [Google Scholar]
  • [146].King TJ, Gurley KE, Prunty J, Shin JL, Kemp CJ, Lampe PD. Deficiency in the gap junction protein connexin32 alters p27Kip1 tumor suppression and MAPK activation in a tissue- specific manner. Oncogene. 2005;24:1718–1726. doi: 10.1038/sj.onc.1208355. [DOI] [PubMed] [Google Scholar]
  • [147].Hokaiwado N, Asamoto M, Ogawa K, Shirai T. Transgenic disruption of gap junctional intercellular communication enhances early but not late stage hepatocarcinogenesis in the rat. Toxicol Pathol. 2005;33:695–701. doi: 10.1080/01926230500330313. [DOI] [PubMed] [Google Scholar]
  • [148].Hokaiwado N, Asamoto M, Futakuchi M, Ogawa K, Takahashi S, Shirai T. Both early and late stages of hepatocarcinogenesis are enhanced in Cx32 dominant negative mutant transgenic rats with disrupted gap junctional intercellular communication. J Membr Biol. 2007;218:101–106. doi: 10.1007/s00232-007-9053-9. [DOI] [PubMed] [Google Scholar]
  • [149].Gotow T, Shiozaki M, Higashi T, Yoshimura K, Shibata M, Kominami E et al. Hepatic gap junctions in the hepatocarcinogen-resistant DRH rat. Histochem Cell Biol. 2008;130:583–594. doi: 10.1007/s00418-008-0473-0. [DOI] [PubMed] [Google Scholar]
  • [150].D’hondt C, Iyyathurai J, Himpens B, Leybaert L, Bultynck G. Cx43-hemichannel function and regulation in physiology and pathophysiology: insights from the bovine corneal endothelial cell system and beyond. Front Physiol. 2014;5:348. doi: 10.3389/fphys.2014.00348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Maes M, Yanguas SC, Willebrords J, Cogliati B, Vinken M. Connexin and pannexin signaling in gastrointestinal and liver disease. Transl Res. 2015;166:332–343. doi: 10.1016/j.trsl.2015.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Dahl G, Keane RW. Pannexin: From discovery to bedside in 11 +/- 4 years? Brain Res. 2012;1487:150–159. doi: 10.1016/j.brainres.2012.04.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Penuela S, Harland L, Simek J, Laird DW. Pannexin channels and their links to human disease. Biochem J. 2014;461:371–381. doi: 10.1042/BJ20140447. [DOI] [PubMed] [Google Scholar]
  • [154].Bruzzone R, Hormuzdi SG, Barbe MT, Herb A, Monyer H. Pannexins, a family of gap junction proteins expressed in brain. Proc Natl Acad Sci U S A. 2003;100:13644–13649. doi: 10.1073/pnas.2233464100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Csak T, Ganz M, Pespisa J, Kodys K, Dolganiuc A, Szabo G. Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells. Hepatology. 2011;54:133–144. doi: 10.1002/hep.24341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Ganz M, Csak T, Nath B, Szabo G. Lipopolysaccharide induces and activates the Nalp3 inflammasome in the liver. World J Gastroenterol. 2011;17:4772–4778. doi: 10.3748/wjg.v17.i43.4772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Kim HY, Kim SJ, Lee SM. Activation of NLRP3 and AIM2 inflammasomes in Kupffer cells in hepatic ischemia/reperfusion. FEBS J. 2015;282:259–270. doi: 10.1111/febs.13123. [DOI] [PubMed] [Google Scholar]
  • [158].Xiao F, Waldrop SL, Khimji AK, Kilic G. Pannexin1 contributes to pathophysiological ATP release in lipoapoptosis induced by saturated free fatty acids in liver cells. Am J Physiol Cell Physiol. 2012;303:C1034–1044. doi: 10.1152/ajpcell.00175.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Sáez PJ, Shoji KF, Aguirre A, Sáez JC. Regulation of hemichannels and gap junction channels by cytokines in antigen- presenting cells. Mediators Inflamm. 2014;2014:742734. doi: 10.1155/2014/742734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Ruch RJ, Klaunig JE. Effects of tumor promoters, genotoxic carcinogens and hepatocytotoxins on mouse hepatocyte inter-cellular communication. Cell Biol Toxicol. 1986;2:469–483. doi: 10.1007/BF00117849. [DOI] [PubMed] [Google Scholar]
  • [161].Budunova IV, Williams GM. Cell culture assays for chemicals with tumor-promoting or tumor-inhibiting activity based on the modulation of intercellular communication. Cell Biol Toxicol. 1994;10:71–116. doi: 10.1007/BF00756491. [DOI] [PubMed] [Google Scholar]
  • [162].Mesnil M, Krutovskikh V, Omori Y, Yamasaki H. Role of blocked gap junctional intercellular communication in non- genotoxic carcinogenesis. Toxicol Lett. 1995;82-83:701–706. doi: 10.1016/0378-4274(95)03588-5. [DOI] [PubMed] [Google Scholar]
  • [163].Combes RD. The use of structure-activity relationships and markers of cell toxicity to detect non-genotoxic carcinogens. Toxicol In Vitro. 2000;14:387–399. doi: 10.1016/s0887-2333(00)00026-6. [DOI] [PubMed] [Google Scholar]
  • [164].Kuraoka A, Iida H, Hatae T, Shibata Y, Itoh M, Kurita T. Localization of gap junction proteins, connexins 32 and 26, in rat and guinea pig liver as revealed by quick-freeze, deep-etch immunoelectron microscopy. J Histochem Cytochem. 1993;41:971–980. doi: 10.1177/41.7.8390496. [DOI] [PubMed] [Google Scholar]
  • [165].Nicholson B, Dermietzel R, Teplow D, Traub O, Willecke K, Revel JP. Two homologous protein components of hepatic gap junctions. Nature. 1987;329:732–734. doi: 10.1038/329732a0. [DOI] [PubMed] [Google Scholar]
  • [166].Zhang JT, Nicholson BJ. Sequence and tissue distribution of a second protein of hepatic gap junctions, Cx26, as deduced from its cDNA. J Cell Biol. 1989;109:3391–3401. doi: 10.1083/jcb.109.6.3391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Fowler SL, Akins M, Zhou H, Figeys D, Bennett SA. The liver connexin32 interactome is a novel plasma membrane-mitochondrial signaling nexus. J Proteome Res. 2013;12:2597–2610. doi: 10.1021/pr301166p. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Balasubramaniyan V, Dhar DK, Warner AE, Vivien Li WY, Amiri AF, Bright B et al. Importance of Connexin-43 based gap junction in cirrhosis and acute-on-chronic liver failure. J Hepatol. 2013;58:1194–1200. doi: 10.1016/j.jhep.2013.01.023. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Clinical and Translational Research are provided here courtesy of Whioce Publishing Pte. Ltd.

RESOURCES